1
|
Moss KR, Saxena S. Schwann Cells in Neuromuscular Disorders: A Spotlight on Amyotrophic Lateral Sclerosis. Cells 2025; 14:47. [PMID: 39791748 PMCID: PMC11719703 DOI: 10.3390/cells14010047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/23/2024] [Accepted: 01/01/2025] [Indexed: 01/12/2025] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a complex neurodegenerative disease primarily affecting motor neurons, leading to progressive muscle atrophy and paralysis. This review explores the role of Schwann cells in ALS pathogenesis, highlighting their influence on disease progression through mechanisms involving demyelination, neuroinflammation, and impaired synaptic function. While Schwann cells have been traditionally viewed as peripheral supportive cells, especially in motor neuron disease, recent evidence indicates that they play a significant role in ALS by impacting motor neuron survival and plasticity, influencing inflammatory responses, and altering myelination processes. Furthermore, advancements in understanding Schwann cell pathology in ALS combined with lessons learned from studying Charcot-Marie-Tooth disease Type 1 (CMT1) suggest potential therapeutic strategies targeting these cells may support nerve repair and slow disease progression. Overall, this review aims to provide comprehensive insights into Schwann cell classification, physiology, and function, underscoring the critical pathological contributions of Schwann cells in ALS and suggests new avenues for targeted therapeutic interventions aimed at modulating Schwann cell function in ALS.
Collapse
Affiliation(s)
- Kathryn R. Moss
- Department of Physical Medicine and Rehabilitation, University of Missouri School of Medicine, Columbia, MO 65211, USA
- NextGen Precision Health, University of Missouri, Columbia, MO 65211, USA
| | - Smita Saxena
- Department of Physical Medicine and Rehabilitation, University of Missouri School of Medicine, Columbia, MO 65211, USA
- NextGen Precision Health, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
2
|
Lavriha P, Han Y, Ding X, Schuster D, Qi C, Vaithia A, Picotti P, Korkhov VM. Mechanism of connexin channel inhibition by mefloquine and 2-aminoethoxydiphenyl borate. PLoS One 2024; 19:e0315510. [PMID: 39739741 DOI: 10.1371/journal.pone.0315510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/26/2024] [Indexed: 01/02/2025] Open
Abstract
Gap junction intercellular communication (GJIC) between two adjacent cells involves direct exchange of cytosolic ions and small molecules via connexin gap junction channels (GJCs). Connexin GJCs have emerged as drug targets, with small molecule connexin inhibitors considered a viable therapeutic strategy in several diseases. The molecular mechanisms of GJC inhibition by known small molecule connexin inhibitors remain unknown, preventing the development of more potent and connexin-specific therapeutics. Here we show that two GJC inhibitors, mefloquine (MFQ) and 2-aminoethoxydiphenyl borate (2APB) bind to Cx32 and block dye permeation across Cx32 hemichannels (HCs) and GJCs. Cryo-EM analysis shows that 2APB binds to "site A", close to the N-terminal gating helix of Cx32 GJC, restricting the entrance to the channel pore. In contrast, MFQ binds to a distinct "site M", deeply buried within the pore. MFQ binding to this site modifies the electrostatic properties of Cx32 pore. Mutagenesis of V37, a key residue located in the site M, renders Cx32 HCs and GJCs insensitive to MFQ-mediated inhibition. Moreover, our cryo-EM analysis, mutagenesis and activity assays show that MFQ targets the M site in Cx43 GJC similarly to Cx32. Taken together, our results point to a conserved inhibitor binding site in connexin channels, opening a new route for development of specific drugs targeting connexins.
Collapse
Affiliation(s)
- Pia Lavriha
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zürich, Switzerland
| | - Yufei Han
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zürich, Switzerland
| | - Xinyue Ding
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zürich, Switzerland
| | - Dina Schuster
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
- Institute of Molecular Systems Biology, ETH Zurich, Zürich, Switzerland
| | - Chao Qi
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zürich, Switzerland
- Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Anand Vaithia
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Paola Picotti
- Institute of Molecular Systems Biology, ETH Zurich, Zürich, Switzerland
| | - Volodymyr M Korkhov
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zürich, Switzerland
| |
Collapse
|
3
|
Tettey-Matey A, Donati V, Cimmino C, Di Pietro C, Buratto D, Panarelli M, Reale A, Calistri A, Fornaini MV, Zhou R, Yang G, Zonta F, Marazziti D, Mammano F. A fully human IgG1 antibody targeting connexin 32 extracellular domain blocks CMTX1 hemichannel dysfunction in an in vitro model. Cell Commun Signal 2024; 22:589. [PMID: 39639332 PMCID: PMC11619691 DOI: 10.1186/s12964-024-01969-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
Connexins (Cxs) are fundamental in cell-cell communication, functioning as gap junction channels (GJCs) that facilitate solute exchange between adjacent cells and as hemichannels (HCs) that mediate solute exchange between the cytoplasm and the extracellular environment. Mutations in the GJB1 gene, which encodes Cx32, lead to X-linked Charcot-Marie-Tooth type 1 (CMTX1), a rare hereditary demyelinating disorder of the peripheral nervous system (PNS) without an effective cure or treatment. In Schwann cells, Cx32 HCs are thought to play a role in myelination by enhancing intracellular and intercellular Ca2+ signaling, which is crucial for proper PNS myelination. Single-point mutations (p.S85C, p.D178Y, p.F235C) generate pathological Cx32 HCs characterized by increased permeability ("leaky") or excessive activity ("hyperactive").We investigated the effects of abEC1.1-hIgG1, a fully human immunoglobulin G1 (hIgG1) monoclonal antibody, on wild-type (WT) and mutant Cx32D178Y HCs. Using HeLa DH cells conditionally co-expressing Cx and a genetically encoded Ca2+ biosensor (GCaMP6s), we demonstrated that mutant HCs facilitated 58% greater Ca2+ uptake in response to elevated extracellular Ca2+ concentrations ([Ca2+]ex) compared to WT HCs. abEC1.1-hIgG1 dose-dependently inhibited Ca2+ uptake, achieving a 50% inhibitory concentration (EC50) of ~ 10 nM for WT HCs and ~ 80 nM for mutant HCs. Additionally, the antibody suppressed DAPI uptake and ATP release. An atomistic computational model revealed that serine 56 (S56) of the antibody interacts with aspartate 178 (D178) of WT Cx32 HCs, contributing to binding affinity. Despite the p.D178Y mutation weakening this interaction, the antibody maintained binding to the mutant HC epitope at sub-micromolar concentrations.In conclusion, our study shows that abEC1.1-hIgG1 effectively inhibits both WT and mutant Cx32 HCs, highlighting its potential as a therapeutic approach for CMTX1. These findings expand the antibody's applicability for treating diseases associated with Cx HCs and inform the rational design of next-generation antibodies with enhanced affinity and efficacy against mutant HCs.
Collapse
Affiliation(s)
- Abraham Tettey-Matey
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, 00015, Italy
- Present Address, CNR Institute of Biophysics, Genoa, 16149, Italy
| | - Viola Donati
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, 00015, Italy
- Department of Biomedical Sciences, University of Padua, Padua, 35131, Italy
| | - Chiara Cimmino
- CNR Institute of Endocrinology and Experimental Oncology "G. Salvatore", Naples, 80131, Italy
- Present Address: Interdisciplinary Research Centre On Biomaterials, University of Naples Federico II, Naples, 80125, Italy
| | - Chiara Di Pietro
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, 00015, Italy
| | - Damiano Buratto
- Institute of Quantitative Biology, College of Life Science, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | | | - Alberto Reale
- Department of Molecular Medicine, University of Padua, Padua, 35131, Italy
| | - Arianna Calistri
- Department of Molecular Medicine, University of Padua, Padua, 35131, Italy
| | | | - Ruhong Zhou
- Institute of Quantitative Biology, College of Life Science, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Guang Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, P. R. China
| | - Francesco Zonta
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, P. R. China.
| | - Daniela Marazziti
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, 00015, Italy.
| | - Fabio Mammano
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, 00015, Italy.
- Department of Physics and Astronomy "G. Galilei", University of Padua, Padua, 35131, Italy.
| |
Collapse
|
4
|
Bayraktar E, Lopez-Pigozzi D, Bortolozzi M. Calcium Regulation of Connexin Hemichannels. Int J Mol Sci 2024; 25:6594. [PMID: 38928300 PMCID: PMC11204158 DOI: 10.3390/ijms25126594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Connexin hemichannels (HCs) expressed at the plasma membrane of mammalian cells are of paramount importance for intercellular communication. In physiological conditions, HCs can form gap junction (GJ) channels, providing a direct diffusive path between neighbouring cells. In addition, unpaired HCs provide conduits for the exchange of solutes between the cytoplasm and the extracellular milieu, including messenger molecules involved in paracrine signalling. The synergistic action of membrane potential and Ca2+ ions controls the gating of the large and relatively unselective pore of connexin HCs. The four orders of magnitude difference in gating sensitivity to the extracellular ([Ca2+]e) and the cytosolic ([Ca2+]c) Ca2+ concentrations suggests that at least two different Ca2+ sensors may exist. While [Ca2+]e acts as a spatial modulator of the HC opening, which is most likely dependent on the cell layer, compartment, and organ, [Ca2+]c triggers HC opening and the release of extracellular bursts of messenger molecules. Such molecules include ATP, cAMP, glutamate, NAD+, glutathione, D-serine, and prostaglandins. Lost or abnormal HC regulation by Ca2+ has been associated with several diseases, including deafness, keratitis ichthyosis, palmoplantar keratoderma, Charcot-Marie-Tooth neuropathy, oculodentodigital dysplasia, and congenital cataracts. The fact that both an increased and a decreased Ca2+ sensitivity has been linked to pathological conditions suggests that Ca2+ in healthy cells finely tunes the normal HC function. Overall, further investigation is needed to clarify the structural and chemical modifications of connexin HCs during [Ca2+]e and [Ca2+]c variations. A molecular model that accounts for changes in both Ca2+ and the transmembrane voltage will undoubtedly enhance our interpretation of the experimental results and pave the way for developing therapeutic compounds targeting specific HC dysfunctions.
Collapse
Affiliation(s)
- Erva Bayraktar
- Veneto Institute of Molecular Medicine (VIMM), Via Orus 2, 35129 Padova, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padua, Via Marzolo 8, 35131 Padova, Italy
| | - Diego Lopez-Pigozzi
- Veneto Institute of Molecular Medicine (VIMM), Via Orus 2, 35129 Padova, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padua, Via Marzolo 8, 35131 Padova, Italy
| | - Mario Bortolozzi
- Veneto Institute of Molecular Medicine (VIMM), Via Orus 2, 35129 Padova, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padua, Via Marzolo 8, 35131 Padova, Italy
- Institute of Endocrinology and Oncology “Gaetano Salvatore” (IEOS-CNR), Via Pietro Castellino 111, 80131 Napoli, Italy
| |
Collapse
|
5
|
Caballé RB, Bortolozzi M. New perspectives for gene therapy of the X-linked form of Charcot-Marie-Tooth disease. Mol Ther Methods Clin Dev 2024; 32:101184. [PMID: 38292668 PMCID: PMC10827554 DOI: 10.1016/j.omtm.2023.101184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Affiliation(s)
- Rafael Balada Caballé
- University of Padua, Department of Physics and Astronomy “G. Galilei”, Padua, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Mario Bortolozzi
- University of Padua, Department of Physics and Astronomy “G. Galilei”, Padua, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
- Padova Neuroscience Center (PNC), Padua, Italy
| |
Collapse
|
6
|
Zhan F, Tian W, Cao Y, Wu J, Ni R, Liu T, Yuan Y, Luan X, Cao L. Episodic Neurological Dysfunction in X-Linked Charcot-Marie-Tooth Disease: Expansion of the Phenotypic and Genetic Spectrum. J Clin Neurol 2024; 20:59-66. [PMID: 38179633 PMCID: PMC10782082 DOI: 10.3988/jcn.2023.0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/10/2023] [Accepted: 05/30/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND AND PURPOSE X-linked Charcot-Marie-Tooth disease type 1 (CMTX1) is characterized by peripheral neuropathy with or without episodic neurological dysfunction. We performed clinical, neuropathological, and genetic investigations of a series of patients with mutations of the gap-junction beta-1 gene (GJB1) to extend the phenotypic and genetic description of CMTX1. METHODS Detailed clinical evaluations, sural nerve biopsy, and genetic analysis were applied to patients with CMTX1. RESULTS We collected 27 patients with CMTX1 with GJB1 mutations from 14 unrelated families. The age at onset (AAO) was 20.9±12.2 years (mean±standard deviation; range, 2-45 years). Walking difficulties, weakness in the legs, and pes cavus were common initial symptoms. Compared with female patients, males tended to have a younger AAO (males vs. females=15.4±9.6 vs. 32.0±8.8 years, p=0.002), a longer disease course (16.8±16.1 vs. 5.5±3.8 years, p=0.034), and more-severe electrophysiological results. Besides peripheral neuropathy, six of the patients had special episodic central nervous system (CNS) evidence from symptoms, signs, and/or reversible white-matter lesions. Neuropathology revealed the loss of large myelinated fibers, increased number of regenerated axon clusters with abnormally thin myelin sheaths, and excessively folded myelin. Genetic analysis identified 14 GJB1 variants, 6 of which were novel. CONCLUSIONS These findings expand the phenotypic and genetic spectrum of CMTX1. Although CMTX1 was found to have high phenotypic and CNS involvement variabilities, detailed neurological examinations and nerve conduction studies will provide critical clues for accurate diagnoses. Further exploration of the underlying mechanisms of connexin 32 involvement in neuropathy or CNS dysfunction is warranted to develop promising therapies.
Collapse
Affiliation(s)
- Feixia Zhan
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wotu Tian
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuwen Cao
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingying Wu
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruilong Ni
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Taotao Liu
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Xinghua Luan
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Li Cao
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Medicine, Anhui University of Science and Technology, Huainan, China.
| |
Collapse
|
7
|
Nozaki I, Hashiguchi A, Takashima H, Yamashita Y, Higashide T, Iwasa K, Ono K. Charcot-Marie-Tooth Disease with a Novel Variant in Gap Junction Protein Beta 1 Presenting with Visual Field Defects. Intern Med 2023; 62:3033-3036. [PMID: 36792185 PMCID: PMC10641200 DOI: 10.2169/internalmedicine.1403-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/10/2023] [Indexed: 02/16/2023] Open
Abstract
Pathogenic variants in Gap Junction Protein Beta 1 (GJB1) cause X-linked Charcot-Marie-Tooth (CMT) disease type 1 (CMTX1), which is a common hereditary motor and sensory neuropathy. A 45-year-old man presented with progressive muscle weakness, atrophy, sensory disturbance of all limbs from childhood, and visual field defects in both eyes at 40 years old. A segregation analysis revealed a novel variant, c.173C>A (p.P58H), in the GJB1 gene. Patients with variants at codon 58 in GJB1 showed clinically varied phenotypes, ranging from demyelinating neuropathy to cerebellar ataxia. This patient may represent one of the various clinical phenotypes of GJB1 variants.
Collapse
Affiliation(s)
- Ichiro Nozaki
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Japan
| | - Akihiro Hashiguchi
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Japan
| | - Hiroshi Takashima
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Japan
| | - Yoko Yamashita
- Department of Opthalmology and Visual Science, Kanazawa University Graduate School of Medical Sciences, Japan
| | - Tomomi Higashide
- Department of Opthalmology and Visual Science, Kanazawa University Graduate School of Medical Sciences, Japan
| | - Kazuo Iwasa
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Japan
- Department of Health and Medical Sciences, Ishikawa Prefectural Nursing University, Japan
| | - Kenjiro Ono
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Japan
| |
Collapse
|
8
|
Qi C, Lavriha P, Bayraktar E, Vaithia A, Schuster D, Pannella M, Sala V, Picotti P, Bortolozzi M, Korkhov VM. Structures of wild-type and selected CMT1X mutant connexin 32 gap junction channels and hemichannels. SCIENCE ADVANCES 2023; 9:eadh4890. [PMID: 37647412 PMCID: PMC10468125 DOI: 10.1126/sciadv.adh4890] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/27/2023] [Indexed: 09/01/2023]
Abstract
In myelinating Schwann cells, connection between myelin layers is mediated by gap junction channels (GJCs) formed by docked connexin 32 (Cx32) hemichannels (HCs). Mutations in Cx32 cause the X-linked Charcot-Marie-Tooth disease (CMT1X), a degenerative neuropathy without a cure. A molecular link between Cx32 dysfunction and CMT1X pathogenesis is still missing. Here, we describe the high-resolution cryo-electron cryo-myography (cryo-EM) structures of the Cx32 GJC and HC, along with two CMT1X-linked mutants, W3S and R22G. While the structures of wild-type and mutant GJCs are virtually identical, the HCs show a major difference: In the W3S and R22G mutant HCs, the amino-terminal gating helix partially occludes the pore, consistent with a diminished HC activity. Our results suggest that HC dysfunction may be involved in the pathogenesis of CMT1X.
Collapse
Affiliation(s)
- Chao Qi
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Pia Lavriha
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Erva Bayraktar
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padua, Padua, Italy
| | - Anand Vaithia
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Dina Schuster
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Micaela Pannella
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padua, Padua, Italy
| | - Valentina Sala
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Paola Picotti
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Mario Bortolozzi
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padua, Padua, Italy
| | - Volodymyr M. Korkhov
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| |
Collapse
|
9
|
Tadenev ALD, Hatton CL, Pattavina B, Mullins T, Schneider R, Bogdanik LP, Burgess RW. Two new mouse models of Gjb1-associated Charcot-Marie-Tooth disease type 1X. J Peripher Nerv Syst 2023; 28:317-328. [PMID: 37551045 DOI: 10.1111/jns.12588] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/25/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND Charcot-Marie-Tooth disease type 1X is caused by mutations in GJB1, which is the second most common gene associated with inherited peripheral neuropathy. The GJB1 gene encodes connexin 32 (CX32), a gap junction protein expressed in myelinating glial cells. The gene is X-linked, and the mutations cause a loss of function. AIMS A large number of disease-associated variants have been identified, and many result in mistrafficking and mislocalization of the protein. An existing knockout mouse lacking Gjb1 expression provides a valid animal model of CMT1X, but the complete lack of protein may not fully recapitulate the disease mechanisms caused by aberrant CX32 proteins. To better represent the spectrum of human CMT1X-associated mutations, we have generated a new Gjb1 knockin mouse model. METHODS CRISPR/Cas9 genome editing was used to produce mice carrying the R15Q mutation in Gjb1. In addition, we identified a second allele with an early frame shift mutation in codon 7 (del2). Mice were analyzed using clinically relevant molecular, histological, neurophysiological, and behavioral assays. RESULTS Both alleles produce protein detectable by immunofluorescence in Schwann cells, with some protein properly localizing to nodes of Ranvier. However, both alleles also result in peripheral neuropathy with thinly myelinated and demyelinated axons, as well as degenerating and regenerating axons, predominantly in distal motor nerves. Nerve conduction velocities were only mildly reduced at later ages and compound muscle action potential amplitudes were not reduced. Levels of neurofilament light chain in plasma were elevated in both alleles. The del2 mice have an onset at ~3 months of age, whereas the R15Q mice had a later onset at 5-6 months of age, suggesting a milder loss of function. Both alleles performed comparably to wild type littermates in accelerating rotarod and grip strength tests of neuromuscular performance. INTERPRETATION We have generated and characterized two new mouse models of CMT1X that will be useful for future mechanistic and preclinical studies.
Collapse
Affiliation(s)
| | - C L Hatton
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | - B Pattavina
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | - T Mullins
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | - R Schneider
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | | | | |
Collapse
|
10
|
Ca 2+-Dependent and -Independent Calmodulin Binding to the Cytoplasmic Loop of Gap Junction Connexins. Int J Mol Sci 2023; 24:ijms24044153. [PMID: 36835569 PMCID: PMC9961272 DOI: 10.3390/ijms24044153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Ca2+/calmodulin (Ca2+/CaM) interaction with connexins (Cx) is well-established; however, the mechanistic basis of regulation of gap junction function by Ca2+/CaM is not fully understood. Ca2+/CaM is predicted to bind to a domain in the C-terminal portion of the intracellular loop (CL2) in the vast majority of Cx isoforms and for a number of Cx-s this prediction has proved correct. In this study, we investigate and characterise both Ca2+/CaM and apo-CaM binding to selected representatives of each of the α, β and γ connexin family to develop a better mechanistic understanding of CaM effects on gap junction function. The affinity and kinetics Ca2+/CaM and apo-CaM interactions of CL2 peptides of β-Cx32, γ-Cx35, α-Cx43, α-Cx45 and α-Cx57 were investigated. All five Cx CL2 peptides were found to have high affinity for Ca2+/CaM with dissociation constants (Kd(+Ca)) from 20 to 150 nM. The limiting rate of binding and the rates of dissociation covered a broad range. In addition, we obtained evidence for high affinity Ca2+-independent interaction of all five peptides with CaM, consistent with CaM remaining anchored to gap junctions in resting cells. However, for the α-Cx45 and α-Cx57 CL2 peptides, Ca2+-dependent association at resting [Ca2+] of 50-100 nM is indicated in these complexes as one of the CaM Ca2+ binding sites displays high affinity with Kd of 70 and 30 nM for Ca2+, respectively. Furthermore, complex conformational changes were observed in peptide-apo-CaM complexes with the structure of CaM compacted or stretched by the peptide in a concentration dependent manner suggesting that the CL2 domain may undergo helix-to-coil transition and/or forms bundles, which may be relevant in the hexameric gap junction. We demonstrate inhibition of gap junction permeability by Ca2+/CaM in a dose dependent manner, further cementing Ca2+/CaM as a regulator of gap junction function. The motion of a stretched CaM-CL2 complex compacting upon Ca2+ binding may bring about the Ca2+/CaM block of the gap junction pore by a push and pull action on the CL2 C-terminal hydrophobic residues of transmembrane domain 3 (TM3) in and out of the membrane.
Collapse
|
11
|
Kaur S, Zhang X, Patel S, Rodriguez YA, Luther KJ, Alghafli G, Lang RM, Abrams CK, Dobrowsky RT. Pharmacologic Targeting of the C-Terminus of Heat Shock Protein 90 Improves Neuromuscular Function in Animal Models of Charcot Marie Tooth X1 Disease. ACS Pharmacol Transl Sci 2023; 6:306-319. [PMID: 36798471 PMCID: PMC9926526 DOI: 10.1021/acsptsci.2c00223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Indexed: 01/22/2023]
Abstract
Charcot-Marie-Tooth X1 (CMTX1) disease is an inherited peripheral neuropathy that arises from loss-of-function mutations in the protein connexin 32 (Cx32). CMTX1 currently lacks a pharmacologic approach toward disease management, and we have previously shown that modulating the expression of molecular chaperones using novologue therapy may provide a viable disease-modifying approach to treat metabolic and demyelinating neuropathies. Cemdomespib is an orally bioavailable novologue that manifests neuroprotective activity by modulating the expression of heat shock protein 70 (Hsp70). We examined if 1 to 5 months of daily cemdomespib therapy may improve neuropathic symptoms in three mouse models of CMTX1 (Cx32 deficient (Cx32def), T55I-Cx32def, and R75W-Cx32 mice). Daily drug therapy significantly improved motor nerve conduction velocity (MNCV) and grip strength in all three models, but the compound muscle action potential was only improved in Cx32def mice. Drug efficacy required Hsp70 as improvements in MNCV, and the grip strength was abrogated in Cx32def × Hsp70 knockout mice. Five months of novologue therapy was associated with improved neuromuscular junction morphology, femoral motor nerve myelination, reduction in foamy macrophages, and a decrease in Schwann cell c-jun levels. To determine if c-jun may be downstream of Hsp70 and necessary for drug efficacy, c-jun expression was specifically deleted in Schwann cells of Cx32def mice. While the deletion of c-jun worsened the neuropathy, cemdomespib therapy remained effective in improving MNCV and grip strength. Our data show that cemdomespib therapy improves CMTX1-linked neuropathy in an Hsp70-dependent but a c-jun-independent manner and without regard to the nature of the underlying Cx32 mutation.
Collapse
Affiliation(s)
- Sukhmanjit Kaur
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| | - Xinyue Zhang
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| | - Sugandha Patel
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| | - Yssa A. Rodriguez
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| | - Kylie J. Luther
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| | - Ghufran Alghafli
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| | - Ryan M. Lang
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| | - Charles K. Abrams
- Department
of Neurology and Rehabilitation and Biomedical Engineering, College
of Medicine, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Rick T. Dobrowsky
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
12
|
Beloribi-Djefaflia S, Attarian S. Treatment of Charcot-Marie-Tooth neuropathies. Rev Neurol (Paris) 2023; 179:35-48. [PMID: 36588067 DOI: 10.1016/j.neurol.2022.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 12/31/2022]
Abstract
Charcot-Marie-Tooth (CMT) is a heterogeneous group of inherited neuropathies that affect the peripheral nerves and slowly cause progressive disability. Currently, there is no effective therapy. Patients' management is based on rehabilitation and occupational therapy, fatigue, and pain treatment with regular follow-up according to the severity of the disease. In the last three decades, much progress has been made to identify mutations involved in the different types of CMT, decipher the pathophysiology of the disease, and identify key genes and pathways that could be targeted to propose new therapeutic strategies. Genetic therapy is one of the fields of interest to silence genes such as PMP22 in CMT1A or to express GJB1 in CMT1X. Among the most promising molecules, inhibitors of the NRG-1 axis and modulators of UPR or the HDACs enzyme family could be used in different types of CMT.
Collapse
Affiliation(s)
- S Beloribi-Djefaflia
- Reference center for neuromuscular disorders and ALS, AP-HM, CHU La Timone, Marseille, France
| | - S Attarian
- Reference center for neuromuscular disorders and ALS, AP-HM, CHU La Timone, Marseille, France; FILNEMUS, European Reference Network for Rare Diseases (ERN), Marseille, France; Medical Genetics, Aix Marseille Université-Inserm UMR_1251, 13005 Marseille, France.
| |
Collapse
|
13
|
Chu F, Xu J, Wang Y, Li Y, Wang Y, Liu Z, Li C. Novel mutations in GJB1 trigger intracellular aggregation and stress granule formation in X-linked Charcot-Marie-Tooth Disease. Front Neurosci 2022; 16:972288. [PMID: 36225735 PMCID: PMC9548587 DOI: 10.3389/fnins.2022.972288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
X-linked Charcot-Marie-Tooth Disease type 1(CMT1X) is the second most common form of inherited peripheral neuropathy that is caused by mutations in the gap junction beta-1 (GJB1) gene. Using targeted exome-sequencing, we investigated four CMT families from central-southern China and identified two novel missense variants (p.F31S and p.W44G) and two previously reported variants (p.R220Pfs*23 and p.R164Q) of GJB1. All four probands presented typical early-onset peripheral neuropathy, of which the R220Pfs*23 carrier also had neurologic manifestations in the central nervous system. We then constructed GJB1 expression vectors and performed cell biological analysis in vitro. Expression of FLAG-tagged GJB1 at various time points after transfection revealed evident protein aggregation with both wild-type and mutant forms, indicated with immunostaining and immunoblotting. Detergent-based sequential fractionation confirmed that all mutants were higher expressed and more prone to aggregate than the wild-type, whereas the R220Pfs*23 mutant showed the greatest amount of SDS-soluble multimers and monomers among groups. Moreover, intracellular aggregation probably occurs in the endoplasmic reticulum compartment rather than the Golgi apparatus. Gap junction plaques were present in all groups and were only compromised in frameshift mutant. Further evidence reveals significant intracellular stress granule formation induced by mutated GJB1 and impaired cell viability indicative of cytotoxicity of self-aggregates. Together, our findings demonstrate novel GJB1 variants-induced cell stress and dysfunction and provide insights into understanding the pathomechanisms of GJB1-CMTX1 and other related disorders.
Collapse
Affiliation(s)
- Fan Chu
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaming Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingjie Li
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaling Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhijun Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuanzhou Li
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Baracaldo-Santamaría D, Corrales-Hernández MG, Ortiz-Vergara MC, Cormane-Alfaro V, Luque-Bernal RM, Calderon-Ospina CA, Cediel-Becerra JF. Connexins and Pannexins: Important Players in Neurodevelopment, Neurological Diseases, and Potential Therapeutics. Biomedicines 2022; 10:2237. [PMID: 36140338 PMCID: PMC9496069 DOI: 10.3390/biomedicines10092237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Cell-to-cell communication is essential for proper embryonic development and its dysfunction may lead to disease. Recent research has drawn attention to a new group of molecules called connexins (Cxs) and pannexins (Panxs). Cxs have been described for more than forty years as pivotal regulators of embryogenesis; however, the exact mechanism by which they provide this regulation has not been clearly elucidated. Consequently, Cxs and Panxs have been linked to congenital neurodegenerative diseases such as Charcot-Marie-Tooth disease and, more recently, chronic hemichannel opening has been associated with adult neurodegenerative diseases (e.g., Alzheimer's disease). Cell-to-cell communication via gap junctions formed by hexameric assemblies of Cxs, known as connexons, is believed to be a crucial component in developmental regulation. As for Panxs, despite being topologically similar to Cxs, they predominantly seem to form channels connecting the cytoplasm to the extracellular space and, despite recent research into Panx1 (Pannexin 1) expression in different regions of the brain during the embryonic phase, it has been studied to a lesser degree. When it comes to the nervous system, Cxs and Panxs play an important role in early stages of neuronal development with a wide span of action ranging from cellular migration during early stages to neuronal differentiation and system circuitry formation. In this review, we describe the most recent available evidence regarding the molecular and structural aspects of Cx and Panx channels, their role in neurodevelopment, congenital and adult neurological diseases, and finally propose how pharmacological modulation of these channels could modify the pathogenesis of some diseases.
Collapse
Affiliation(s)
- Daniela Baracaldo-Santamaría
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - María Gabriela Corrales-Hernández
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Maria Camila Ortiz-Vergara
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Valeria Cormane-Alfaro
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Ricardo-Miguel Luque-Bernal
- Anatomy and Embriology Units, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Carlos-Alberto Calderon-Ospina
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
- GENIUROS Research Group, Center for Research in Genetics and Genomics (CIGGUR), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Juan-Fernando Cediel-Becerra
- Histology and Embryology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| |
Collapse
|
15
|
Karakaya T, Turkyilmaz A, Sager G, Inan R, Yarali O, Cebi AH, Akin Y. Molecular characterization of Turkish patients with demyelinating Charcot-Marie-Tooth disease. Neurogenetics 2022; 23:213-221. [DOI: 10.1007/s10048-022-00693-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/04/2022] [Indexed: 10/18/2022]
|
16
|
Laird DW, Lampe PD. Cellular mechanisms of connexin-based inherited diseases. Trends Cell Biol 2022; 32:58-69. [PMID: 34429228 PMCID: PMC8688313 DOI: 10.1016/j.tcb.2021.07.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 01/03/2023]
Abstract
The 21-member connexin gene family exhibits distinct tissue expression patterns that can cause a diverse array of over 30 inherited connexin-linked diseases ranging from deafness to skin defects and blindness. Intriguingly, germline mutations can cause disease in one tissue while other tissues that abundantly express the mutant connexin remain disease free, highlighting the importance of the cellular context of mutant expression. Modeling connexin pathologies in genetically modified mice and tissue-relevant cells has informed extensively on no less than a dozen gain- and loss-of-function mechanisms that underpin disease. This review focuses on how a deeper molecular understanding of the over 930 mutations in 11 connexin-encoding genes is foundational for creating a framework for therapeutic interventions.
Collapse
Affiliation(s)
- Dale W. Laird
- Departments of Anatomy and Cell Biology, and Physiology and Pharmacology, University of Western Ontario, London, ON, CANADA
| | - Paul D. Lampe
- Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
17
|
Guo H, Liu Y, Gu J, Luo J, Ma Y, Xiao F. GJB1 mutations c.212T>G and c.311A>C induce apoptosis and inwardly rectifying potassium current changes in X-linked Charcot-Marie-Tooth type 1. Biochem Biophys Res Commun 2021; 582:8-15. [PMID: 34678594 DOI: 10.1016/j.bbrc.2021.10.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/13/2021] [Indexed: 01/29/2023]
Abstract
Gap junction beta 1 (GJB1) is the pathogenic gene of X-linked Charcot-Marie-Tooth type 1 (CMTX1), a rare hereditary sensorimotor neuropathy. However, different mutations of GJB1 result in heterogeneous clinical manifestations with only some mutations leading to central nervous system involvement. We previously reported two GJB1 missense mutations: one novel mutation (c.212T > G) found in a CMTX1 family that only manifested as peripheral neuropathy, and another previously reported mutation GJB1(c.311A > C) leading to involvement of the peripheral nerves and cerebral white matter. However, the mechanism by which GJB1 mutations lead to CMTX1 has not been fully characterized. Here, we generated Schwann cells and primary cultured oligodendrocytes with these two mutations, resulting in the Cx32I71S (GJB1 c.212T > G) and Cx32K104T (GJB1 c.311A > C) mutants, to analyze the pathogenic mechanism using cytology, molecular biology, and electrophysiological methods. Both mutants showed abnormal endoplasmic reticulum aggregation, especially the Cx32K104T mutant, leading to an increase in endoplasmic reticulum stress, resulting in apoptosis. Furthermore, whole-cell patch clamp experiments in oligodendrocytes revealed that the Cx32K104T mutant reduced the cell membrane potential and inwardly rectifying potassium currents, which may be a vital element for central involvement. Therefore, our results may provide a new perspective for understanding the pathogenesis of CMTX1.
Collapse
Affiliation(s)
- Haokun Guo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Yan Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Juan Gu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Jing Luo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Yuanlin Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China.
| | - Fei Xiao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China.
| |
Collapse
|
18
|
Rawat A, Morrison BM. Metabolic Transporters in the Peripheral Nerve-What, Where, and Why? Neurotherapeutics 2021; 18:2185-2199. [PMID: 34773210 PMCID: PMC8804006 DOI: 10.1007/s13311-021-01150-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 12/18/2022] Open
Abstract
Cellular metabolism is critical not only for cell survival, but also for cell fate, function, and intercellular communication. There are several different metabolic transporters expressed in the peripheral nervous system, and they each play important roles in maintaining cellular energy. The major source of energy in the peripheral nervous system is glucose, and glucose transporters 1 and 3 are expressed and allow blood glucose to be imported and utilized by peripheral nerves. There is also increasing evidence that other sources of energy, particularly monocarboxylates such as lactate that are transported primarily by monocarboxylate transporters 1 and 2 in peripheral nerves, can be efficiently utilized by peripheral nerves. Finally, emerging evidence supports an important role for connexins and possibly pannexins in the supply and regulation of metabolic energy. In this review, we will first define these critical metabolic transporter subtypes and then examine their localization in the peripheral nervous system. We will subsequently discuss the evidence, which comes both from experiments in animal models and observations from human diseases, supporting critical roles played by these metabolic transporters in the peripheral nervous system. Despite progress made in understanding the function of these transporters, many questions and some discrepancies remain, and these will also be addressed throughout this review. Peripheral nerve metabolism is fundamentally important and renewed interest in these pathways should help to answer many of these questions and potentially provide new treatments for neurologic diseases that are partly, or completely, caused by disruption of metabolism.
Collapse
Affiliation(s)
- Atul Rawat
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brett M Morrison
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
19
|
Gap Junction Channelopathies and Calmodulinopathies. Do Disease-Causing Calmodulin Mutants Affect Direct Cell-Cell Communication? Int J Mol Sci 2021; 22:ijms22179169. [PMID: 34502077 PMCID: PMC8431743 DOI: 10.3390/ijms22179169] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/19/2021] [Accepted: 08/21/2021] [Indexed: 11/24/2022] Open
Abstract
The cloning of connexins cDNA opened the way to the field of gap junction channelopathies. Thus far, at least 35 genetic diseases, resulting from mutations of 11 different connexin genes, are known to cause numerous structural and functional defects in the central and peripheral nervous system as well as in the heart, skin, eyes, teeth, ears, bone, hair, nails and lymphatic system. While all of these diseases are due to connexin mutations, minimal attention has been paid to the potential diseases of cell–cell communication caused by mutations of Cx-associated molecules. An important Cx accessory protein is calmodulin (CaM), which is the major regulator of gap junction channel gating and a molecule relevant to gap junction formation. Recently, diseases caused by CaM mutations (calmodulinopathies) have been identified, but thus far calmodulinopathy studies have not considered the potential effect of CaM mutations on gap junction function. The major goal of this review is to raise awareness on the likely role of CaM mutations in defects of gap junction mediated cell communication. Our studies have demonstrated that certain CaM mutants affect gap junction channel gating or expression, so it would not be surprising to learn that CaM mutations known to cause diseases also affect cell communication mediated by gap junction channels.
Collapse
|
20
|
Kohrman D, Borges BC, Cassinotti L, Ji L, Corfas G. Axon-glia interactions in the ascending auditory system. Dev Neurobiol 2021; 81:546-567. [PMID: 33561889 PMCID: PMC9004231 DOI: 10.1002/dneu.22813] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/25/2020] [Accepted: 02/05/2021] [Indexed: 11/09/2022]
Abstract
The auditory system detects and encodes sound information with high precision to provide a high-fidelity representation of the environment and communication. In mammals, detection occurs in the peripheral sensory organ (the cochlea) containing specialized mechanosensory cells (hair cells) that initiate the conversion of sound-generated vibrations into action potentials in the auditory nerve. Neural activity in the auditory nerve encodes information regarding the intensity and frequency of sound stimuli, which is transmitted to the auditory cortex through the ascending neural pathways. Glial cells are critical for precise control of neural conduction and synaptic transmission throughout the pathway, allowing for the precise detection of the timing, frequency, and intensity of sound signals, including the sub-millisecond temporal fidelity is necessary for tasks such as sound localization, and in humans, for processing complex sounds including speech and music. In this review, we focus on glia and glia-like cells that interact with hair cells and neurons in the ascending auditory pathway and contribute to the development, maintenance, and modulation of neural circuits and transmission in the auditory system. We also discuss the molecular mechanisms of these interactions, their impact on hearing and on auditory dysfunction associated with pathologies of each cell type.
Collapse
Affiliation(s)
- David Kohrman
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Beatriz C. Borges
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Luis Cassinotti
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Lingchao Ji
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Gabriel Corfas
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| |
Collapse
|
21
|
Moss KR, Bopp TS, Johnson AE, Höke A. New evidence for secondary axonal degeneration in demyelinating neuropathies. Neurosci Lett 2021; 744:135595. [PMID: 33359733 PMCID: PMC7852893 DOI: 10.1016/j.neulet.2020.135595] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/31/2020] [Accepted: 12/19/2020] [Indexed: 12/28/2022]
Abstract
Development of peripheral nervous system (PNS) myelin involves a coordinated series of events between growing axons and the Schwann cell (SC) progenitors that will eventually ensheath them. Myelin sheaths have evolved out of necessity to maintain rapid impulse propagation while accounting for body space constraints. However, myelinating SCs perform additional critical functions that are required to preserve axonal integrity including mitigating energy consumption by establishing the nodal architecture, regulating axon caliber by organizing axonal cytoskeleton networks, providing trophic and potentially metabolic support, possibly supplying genetic translation materials and protecting axons from toxic insults. The intermediate steps between the loss of these functions and the initiation of axon degeneration are unknown but the importance of these processes provides insightful clues. Prevalent demyelinating diseases of the PNS include the inherited neuropathies Charcot-Marie-Tooth Disease, Type 1 (CMT1) and Hereditary Neuropathy with Liability to Pressure Palsies (HNPP) and the inflammatory diseases Acute Inflammatory Demyelinating Polyneuropathy (AIDP) and Chronic Inflammatory Demyelinating Polyneuropathy (CIDP). Secondary axon degeneration is a common feature of demyelinating neuropathies and this process is often correlated with clinical deficits and long-lasting disability in patients. There is abundant electrophysiological and histological evidence for secondary axon degeneration in patients and rodent models of PNS demyelinating diseases. Fully understanding the involvement of secondary axon degeneration in these diseases is essential for expanding our knowledge of disease pathogenesis and prognosis, which will be essential for developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Kathryn R Moss
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Taylor S Bopp
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Anna E Johnson
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Ahmet Höke
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
22
|
Harnessing the therapeutic potential of antibodies targeting connexin hemichannels. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166047. [PMID: 33418036 DOI: 10.1016/j.bbadis.2020.166047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/17/2020] [Accepted: 12/03/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Connexin hemichannels have been implicated in pathology-promoting conditions, including inflammation, numerous widespread human diseases, including cancer and diabetes, and several rare diseases linked to pathological point mutations. METHODS We analysed the literature focusing on antibodies capable of modulating hemichannel function, highlighting generation methods, applications to basic biomedical research and translational potential. RESULTS Anti-hemichannel antibodies generated over the past 3 decades targeted mostly connexin 43, with a focus on cancer treatment. A slow transition from relatively unselective polyclonal antibodies to more selective monoclonal antibodies resulted in few products with interesting characteristics that are under evaluation for clinical trials. Selection of antibodies from combinatorial phage-display libraries, has permitted to engineer a monoclonal antibody that binds to and blocks pathological hemichannels formed by connexin 26, 30 and 32. CONCLUSIONS All known antibodies that modulate connexin hemichannels target the two small extracellular loops of the connexin proteins. The extracellular region of different connexins is highly conserved, and few residues of each connexins are exposed. The search for new antibodies may develop an unprecedented potential for therapeutic applications, as it may benefit tremendously from novel whole-cell screening platforms that permit in situ selection of antibodies against membrane proteins in native state. The demonstrated efficacy of mAbs in reaching and modulating hemichannels in vivo, together with their relative specificity for connexins overlapping epitopes, should hopefully stimulate an interest for widening the scope of anti-hemichannel antibodies. There is no shortage of currently incurable diseases for which therapeutic intervention may benefit from anti-hemichannel antibodies capable of modulating hemichannel function selectively and specifically.
Collapse
|
23
|
Boso F, Taioli F, Cabrini I, Cavallaro T, Fabrizi GM. Aberrant Splicing in GJB1 and the Relevance of 5' UTR in CMTX1 Pathogenesis. Brain Sci 2020; 11:brainsci11010024. [PMID: 33375465 PMCID: PMC7824018 DOI: 10.3390/brainsci11010024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/20/2020] [Accepted: 12/23/2020] [Indexed: 11/16/2022] Open
Abstract
The second most common form of Charcot-Marie-Tooth disease (CMT) follows an X-linked dominant inheritance pattern (CMTX1), referring to mutations in the gap junction protein beta 1 gene (GJB1) that affect connexin 32 protein (Cx32) and its ability to form gap junctions in the myelin sheath of peripheral nerves. Despite the advances of next-generation sequencing (NGS), attention has only recently also focused on noncoding regions. We describe two unrelated families with a c.-17+1G>T transversion in the 5' untranslated region (UTR) of GJB1 that cosegregates with typical features of CMTX1. As suggested by in silico analysis, the mutation affects the regulatory sequence that controls the proper splicing of the intron in the corresponding mRNA. The retention of the intron is also associated with reduced levels of the transcript and the loss of immunofluorescent staining for Cx32 in the nerve biopsy, thus supporting the hypothesis of mRNA instability as a pathogenic mechanism in these families. Therefore, our report corroborates the role of 5' UTR of GJB1 in the pathogenesis of CMTX1 and emphasizes the need to include this region in routine GJB1 screening, as well as in NGS panels.
Collapse
Affiliation(s)
- Federica Boso
- Department of Neurological Sciences, Biomedicine and Movement Sciences, University of Verona, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (F.B.); (F.T.); (I.C.)
- Department of Cellular, Computational and Integrative Biology, University of Trento, Via Sommarive 9, 38123 Povo (Trento), Italy
| | - Federica Taioli
- Department of Neurological Sciences, Biomedicine and Movement Sciences, University of Verona, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (F.B.); (F.T.); (I.C.)
| | - Ilaria Cabrini
- Department of Neurological Sciences, Biomedicine and Movement Sciences, University of Verona, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (F.B.); (F.T.); (I.C.)
| | - Tiziana Cavallaro
- Azienda Ospedaliera Universitaria Integrata Verona—Borgo Roma, Piazzale L.A. Scuro 10, 37134 Verona, Italy;
| | - Gian Maria Fabrizi
- Department of Neurological Sciences, Biomedicine and Movement Sciences, University of Verona, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (F.B.); (F.T.); (I.C.)
- Azienda Ospedaliera Universitaria Integrata Verona—Borgo Roma, Piazzale L.A. Scuro 10, 37134 Verona, Italy;
- Correspondence: ; Tel.: +39-0458124286
| |
Collapse
|
24
|
Liu X, Duan X, Zhang Y, Sun A, Fan D. Cross-Sectional Study in a Large Cohort of Chinese Patients With GJB1 Gene Mutations. Front Neurol 2020; 11:690. [PMID: 32903794 PMCID: PMC7438869 DOI: 10.3389/fneur.2020.00690] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 06/09/2020] [Indexed: 11/24/2022] Open
Abstract
Charcot-Marie-Tooth (CMT) disease is a clinically and genetically heterogeneous group of inherited neuropathies. The GJB1 gene is the pathogenic gene of CMTX1. In this study, we screened a cohort of 465 unrelated Chinese CMT patients from years 2007 to 2019 and 650 controls by direct Sanger sequencing in GJB1 gene or targeted next-generation sequencing (NGS) or whole-exome sequencing (WES). A bidirectional Sanger sequencing would be performed on the 600 bases in the upstream promoter region and 30 bases in the 3′ untranslated region (UTR), if no mutation was found in the coding region of GJB1 of the patient. According to the results, 24 missense mutations, 4 nonsense mutation, 1 entire deletion, 1 intronic mutation, and 4 frameshift mutations in GJB1 were identified. Three of them were novel mutations (c.104 T>C, c.658-659 ins C, and c.811 del G). Moreover, central nervous system involvement was observed in five patients carrying mutations of R15W, V95M, R142W, R164W, and E186K. Our findings expand the mutational spectrum of the GJB1 gene in CMT patients. We also explored the genotype–phenotype correlation according to the collected information in this study. NGS panels for detecting inherited neuropathy should cover the non-coding region of GJB1.
Collapse
Affiliation(s)
- Xiaoxuan Liu
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Xiaohui Duan
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Yingshuang Zhang
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Aping Sun
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
| |
Collapse
|
25
|
Kuang Y, Zorzi V, Buratto D, Ziraldo G, Mazzarda F, Peres C, Nardin C, Salvatore AM, Chiani F, Scavizzi F, Raspa M, Qiang M, Chu Y, Shi X, Li Y, Liu L, Shi Y, Zonta F, Yang G, Lerner RA, Mammano F. A potent antagonist antibody targeting connexin hemichannels alleviates Clouston syndrome symptoms in mutant mice. EBioMedicine 2020; 57:102825. [PMID: 32553574 PMCID: PMC7378960 DOI: 10.1016/j.ebiom.2020.102825] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Numerous currently incurable human diseases have been causally linked to mutations in connexin (Cx) genes. In several instances, pathological mutations generate abnormally active Cx hemichannels, referred to also as "leaky" hemichannels. The goal of this study was to assay the in vivo efficacy of a potent antagonist antibody targeting Cx hemichannels. METHODS We employed the antibody to treat Cx30A88V/A88V adult mutant mice, the only available animal model of Clouston syndrome, a rare orphan disease caused by Cx30 p.A88V leaky hemichannels. To gain mechanistic insight into antibody action, we also performed patch clamp recordings, Ca2+ imaging and ATP release assay in vitro. FINDINGS Two weeks of antibody treatment sufficed to repress cell hyperproliferation in skin and reduce hypertrophic sebaceous glands (SGs) to wild type (wt) levels. These effects were obtained whether mutant mice were treated topically, by application of an antibody cream formulation, or systemically, by intraperitoneal antibody injection. Experiments with mouse primary keratinocytes and HaCaT cells revealed the antibody blocked Ca2+ influx and diminished ATP release through leaky Cx30 p.A88V hemichannels. INTERPRETATION Our results show anti-Cx antibody treatment was effective in vivo and sufficient to counteract the effects of pathological connexin expression in Cx30A88V/A88V mice. In vitro experiments suggest antibodies gained control over leaky hemichannels and contributed to restoring epidermal homeostasis. Therefore, regulating cell physiology by antibodies targeting the extracellular domain of Cxs may enforce an entirely new therapeutic strategy. These findings support the further development of antibodies as drugs to address unmet medical needs for Cx-related diseases. FUND: Fondazione Telethon, GGP19148; University of Padova, SID/BIRD187130; Consiglio Nazionale delle Ricerche, DSB.AD008.370.003\TERABIO-IBCN; National Science Foundation of China, 31770776; Science and Technology Commission of Shanghai Municipality, 16DZ1910200.
Collapse
Affiliation(s)
- Yuanyuan Kuang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, 201210 Shanghai, China; Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031 Shanghai, China; University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Veronica Zorzi
- CNR Institute of Biochemistry and Cell Biology, 00015 Monterotondo, Italy; Institute of Otorhinolaryngology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Damiano Buratto
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Gaia Ziraldo
- CNR Institute of Biochemistry and Cell Biology, 00015 Monterotondo, Italy; Institute of Otorhinolaryngology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Flavia Mazzarda
- CNR Institute of Biochemistry and Cell Biology, 00015 Monterotondo, Italy; Department of Science, Roma3 University, 00146 Rome, Italy
| | - Chiara Peres
- CNR Institute of Biochemistry and Cell Biology, 00015 Monterotondo, Italy; Department of Physics and Astronomy "G. Galilei", University of Padova, 35131 Padova, Italy
| | - Chiara Nardin
- CNR Institute of Biochemistry and Cell Biology, 00015 Monterotondo, Italy; Department of Physics and Astronomy "G. Galilei", University of Padova, 35131 Padova, Italy
| | | | - Francesco Chiani
- CNR Institute of Biochemistry and Cell Biology, 00015 Monterotondo, Italy
| | | | - Marcello Raspa
- CNR Institute of Biochemistry and Cell Biology, 00015 Monterotondo, Italy
| | - Min Qiang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Youjun Chu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Xiaojie Shi
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Yu Li
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, 201210 Shanghai, China; Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031 Shanghai, China; University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Lili Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Yaru Shi
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Francesco Zonta
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Guang Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China.
| | - Richard A Lerner
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; Department of Chemistry, Scripps Research Institute, La Jolla, CA 92037, U.S.A..
| | - Fabio Mammano
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; CNR Institute of Biochemistry and Cell Biology, 00015 Monterotondo, Italy; Department of Physics and Astronomy "G. Galilei", University of Padova, 35131 Padova, Italy.
| |
Collapse
|
26
|
Kober KM, Schumacher M, Conley YP, Topp K, Mazor M, Hammer MJ, Paul SM, Levine JD, Miaskowski C. Signaling pathways and gene co-expression modules associated with cytoskeleton and axon morphology in breast cancer survivors with chronic paclitaxel-induced peripheral neuropathy. Mol Pain 2020; 15:1744806919878088. [PMID: 31486345 PMCID: PMC6755139 DOI: 10.1177/1744806919878088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background The major dose-limiting toxicity of paclitaxel, one of the most commonly used
drugs to treat breast cancer, is peripheral neuropathy (paclitaxel-induced
peripheral neuropathy). Paclitaxel-induced peripheral neuropathy, which
persists into survivorship, has a negative impact on patient’s mood,
functional status, and quality of life. Currently, no interventions are
available to treat paclitaxel-induced peripheral neuropathy. A critical
barrier to the development of efficacious interventions is the lack of
understanding of the mechanisms that underlie paclitaxel-induced peripheral
neuropathy. While data from preclinical studies suggest that disrupting
cytoskeleton- and axon morphology-related processes are a potential
mechanism for paclitaxel-induced peripheral neuropathy, clinical evidence is
limited. The purpose of this study in breast cancer survivors was to
evaluate whether differential gene expression and co-expression patterns in
these pathways are associated with paclitaxel-induced peripheral
neuropathy. Methods Signaling pathways and gene co-expression modules associated with
cytoskeleton and axon morphology were identified between survivors who
received paclitaxel and did (n = 25) or did not (n = 25) develop
paclitaxel-induced peripheral neuropathy. Results Pathway impact analysis identified four significantly perturbed cytoskeleton-
and axon morphology-related signaling pathways. Weighted gene co-expression
network analysis identified three co-expression modules. One module was
associated with paclitaxel-induced peripheral neuropathy group membership.
Functional analysis found that this module was associated with four
signaling pathways and two ontology annotations related to cytoskeleton and
axon morphology. Conclusions This study, which is the first to apply systems biology approaches using
circulating whole blood RNA-seq data in a sample of breast cancer survivors
with and without chronic paclitaxel-induced peripheral neuropathy, provides
molecular evidence that cytoskeleton- and axon morphology-related mechanisms
identified in preclinical models of various types of neuropathic pain
including chemotherapy-induced peripheral neuropathy are found in breast
cancer survivors and suggests pathways and a module of genes for validation
and as potential therapeutic targets.
Collapse
Affiliation(s)
- Kord M Kober
- School of Nursing, University of California, San Francisco, CA, USA
| | - Mark Schumacher
- School of Medicine, University of California, San Francisco, CA, USA
| | - Yvette P Conley
- School of Nursing, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kimberly Topp
- School of Medicine, University of California, San Francisco, CA, USA
| | - Melissa Mazor
- School of Nursing, University of California, San Francisco, CA, USA
| | - Marilynn J Hammer
- Icahn School of Medicine, Mount Sinai Medical Center, New York, NY, USA
| | - Steven M Paul
- School of Nursing, University of California, San Francisco, CA, USA
| | - Jon D Levine
- School of Medicine, University of California, San Francisco, CA, USA
| | | |
Collapse
|
27
|
Michaelidou K, Tsiverdis I, Erimaki S, Papadimitriou D, Amoiridis G, Papadimitriou A, Mitsias P, Zaganas I. Whole exome sequencing establishes diagnosis of Charcot-Marie-Tooth 4J, 1C, and X1 subtypes. Mol Genet Genomic Med 2020; 8:e1141. [PMID: 32022442 PMCID: PMC7196464 DOI: 10.1002/mgg3.1141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 01/01/2020] [Accepted: 01/03/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Charcot-Marie-Tooth (CMT) hereditary polyneuropathies pose a diagnostic challenge. Our aim here is to describe CMT patients diagnosed by whole exome sequencing (WES) following years of fruitless testing. METHODS/RESULTS Three patients with polyneuropathy suspected to be genetic in origin, but not harboring PMP22 gene deletion/duplication, were offered WES. The first patient, a 66-year-old man, had been suffering from progressive weakness and atrophies in the lower and upper extremities for 20 years. Due to ambiguous electrophysiological findings, immune therapies were administered to no avail. Twelve years after PMP22 deletion/duplication testing, WES revealed two pathogenic variants in the FIG4 gene (p.Ile41Thr and p.Phe598fs, respectively), as a cause of CMT 4J. The second patient, a 19-year-old man, had been suffering from hearing and gait impairment since at least his infancy, and recently presented with weakness and dystonia of the lower extremities. In this patient, WES identified the p.Leu122Val LITAF gene variant in heterozygous state, suggesting the diagnosis of CMT 1C, several years after initial genetic analyses. The third patient, a 44-year-old man, presented with progressive weakness and atrophies of the lower and upper extremities since the age of 17 years old. In this patient, WES identified the hemizygous p.Arg164Gln pathogenic variant in the GJB1 gene, establishing the diagnosis of CMT X1, 8 years after testing for PMP22 deletion/duplication. CONCLUSION Novel diagnostic techniques, such as WES, offer the possibility to decipher the cause of CMT subtypes, ending the diagnostic Odyssey of the patients and sparing them from unnecessary and potentially harmful treatments.
Collapse
Affiliation(s)
- Kleita Michaelidou
- Neurogenetics LaboratoryMedical SchoolUniversity of CreteHeraklion, CreteGreece
| | - Ioannis Tsiverdis
- Neurology DepartmentUniversity Hospital of CreteHeraklion, CreteGreece
| | - Sophia Erimaki
- Neurophysiology UnitUniversity Hospital of CreteHeraklion, CreteGreece
| | | | | | | | - Panayiotis Mitsias
- Neurogenetics LaboratoryMedical SchoolUniversity of CreteHeraklion, CreteGreece
- Neurology DepartmentUniversity Hospital of CreteHeraklion, CreteGreece
- Neurophysiology UnitUniversity Hospital of CreteHeraklion, CreteGreece
- Department of NeurologyHenry Ford Hospital/Wayne State UniversityDetroitMIUSA
| | - Ioannis Zaganas
- Neurogenetics LaboratoryMedical SchoolUniversity of CreteHeraklion, CreteGreece
- Neurology DepartmentUniversity Hospital of CreteHeraklion, CreteGreece
| |
Collapse
|
28
|
Sargiannidou I, Kagiava A, Kleopa KA. Gene therapy approaches targeting Schwann cells for demyelinating neuropathies. Brain Res 2020; 1728:146572. [PMID: 31790684 DOI: 10.1016/j.brainres.2019.146572] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/12/2019] [Accepted: 11/26/2019] [Indexed: 11/27/2022]
Abstract
Charcot-Marie-Tooth disease (CMT) encompasses numerous genetically heterogeneous inherited neuropathies, which together are one of the commonest neurogenetic disorders. Axonal CMT types result from mutations in neuronally expressed genes, whereas demyelinating CMT forms mostly result from mutations in genes expressed by myelinating Schwann cells. The demyelinating forms are the most common, and may be caused by dominant mutations and gene dosage effects (as in CMT1), as well as by recessive mutations and loss of function mechanisms (as in CMT4). The discovery of causative genes and increasing insights into molecular mechanisms through the study of experimental disease models has provided the basis for the development of gene therapy approaches. For demyelinating CMT, gene silencing or gene replacement strategies need to be targeted to Schwann cells. Progress in gene replacement for two different CMT forms, including CMT1X caused by GJB1 gene mutations, and CMT4C, caused by SH3TC2 gene mutations, has been made through the use of a myelin-specific promoter to restrict expression in Schwann cells, and by lumbar intrathecal delivery of lentiviral viral vectors to achieve more widespread biodistribution in the peripheral nervous system. This review summarizes the molecular-genetic mechanisms of selected demyelinating CMT neuropathies and the progress made so far, as well as the remaining challenges in the path towards a gene therapy to treat these disorders through the use of optimal gene therapy tools including clinically translatable delivery methods and adeno-associated viral (AAV) vectors.
Collapse
Affiliation(s)
- Irene Sargiannidou
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Alexia Kagiava
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Kleopas A Kleopa
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus; Neurology Clinics, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus.
| |
Collapse
|
29
|
Zhao ZH, Chen ZT, Zhou RL, Wang YZ. A Chinese pedigree with a novel mutation in GJB1 gene and a rare variation in DHTKD1 gene for diverse Charcot‑Marie‑Tooth diseases. Mol Med Rep 2019; 19:4484-4490. [PMID: 30896807 DOI: 10.3892/mmr.2019.10058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/07/2019] [Indexed: 11/05/2022] Open
Abstract
Charcot‑Marie‑Tooth (CMT) disease is a group of motor and sensory neuropathies with a high degree of pathological and genetic heterogenicity. The present study described 2 patients with CMT in a Chinese Han pedigree. The proband exhibited the classic manifestation of CMT with slowly progressing muscular atrophy and weakness. Electrophysiological examination highlighted axonal and demyelinating features. His mother did not have any symptoms, but did exhibit abnormal electrophysiological results. Next‑generation sequencing technology was employed to screen mutations in the genes associated with inherited motor never diseases. A novel mutation, c.528_530delAGT, in the gap junction protein beta 1 (GJB1) gene for CMTX, and a rare variation, c.2369C>T, in the dehydrogenase E1 and transketolase domain containing 1 (DHTKD1) gene for CMT disease type 2Q (CMT2Q), were identified in the proband and his mother. The results were verified by Sanger sequencing. Although the in silico analysis predicted no change in the 3‑dimensional structure, the clinical and electrophysiological presentation in the pedigree and the high evolutionary conservation of the affected amino acid supported the hypothesis that the c.528_530delAGT mutation in the GJB1 gene may be pathogenic in this pedigree. In silico analysis and high evolutionary conservation suggested the pathogenicity of the c.2369C>T mutation in the DHTKD1 gene; however, the clinical and electrophysiological performances of the proband and his mother did not conform to those of CMT2Q caused by the DHTKD1 gene. The present study provided additional information concerning the range of mutations of the GJB1 gene, which facilitated the understanding of the genotype‑phenotype association of CMT.
Collapse
Affiliation(s)
- Zhen-Hua Zhao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Zhi-Ting Chen
- Department of Neurology, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Rui-Ling Zhou
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Yin-Zhou Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
30
|
Cisterna BA, Arroyo P, Puebla C. Role of Connexin-Based Gap Junction Channels in Communication of Myelin Sheath in Schwann Cells. Front Cell Neurosci 2019; 13:69. [PMID: 30881289 PMCID: PMC6405416 DOI: 10.3389/fncel.2019.00069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 02/12/2019] [Indexed: 12/21/2022] Open
Abstract
Peripheral nerves have the capacity to conduct action potentials along great distances and quickly recover following damage which is mainly due to Schwann cells (SCs), the most abundant glial cells of the peripheral nervous system (PNS). SCs wrap around an axonal segment multiple times, forming a myelin sheath, allowing for a significant increase in action potential conduction by insulating the axons. Mature myelin consists of compact and non-compact (or cytoplasmic) myelin zones. Non-compact myelin is found in paranodal loops bordering the nodes of Ranvier, and in the inner and outermost cytoplasmic tongues and is the region in which Schmidt-Lanterman incisures (SLI; continuous spirals of overlapping cytoplasmic expansions within areas of compact myelin) are located. Using different technologies, it was shown that the layers of non-compact myelin could be connected to each other by gap junction channels (GJCs), formed by connexin 32 (Cx32), and their relative abundance allows for the transfer of ions and different small molecules. Likewise, Cx29 is expressed in the innermost layer of the myelin sheath. Here it does not form GJCs but colocalizes with Kv1, which implies that the SCs play an active role in the electrical condition in mammals. The critical role of GJCs in the functioning of myelinating SCs is evident in Charcot-Marie-Tooth disease (CMT), X-linked form 1 (CMTX1), which is caused by mutations in the gap junction protein beta 1 (GJB1) gene that codes for Cx32. Although the management of CMT symptoms is currently supportive, there is a recent method for targeted gene delivery to myelinating cells, which rescues the phenotype in KO-Cx32 mice, a model of CMTX1. In this mini-review article, we discuss the current knowledge on the role of Cxs in myelin-forming SCs and summarize recent discoveries that may become a real treatment possibility for patients with disorders such as CMT.
Collapse
Affiliation(s)
- Bruno A Cisterna
- Escuela de Medicina, Universidad de Talca, Talca, Chile.,Centro para el Desarrollo de la Nanociencia y Nanotecnología (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Pablo Arroyo
- Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos Puebla
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| |
Collapse
|